Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Mol Brain ; 14(1): 149, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34629097

ABSTRACT

The generation of mature synaptic structures using neurons differentiated from human-induced pluripotent stem cells (hiPSC-neurons) is expected to be applied to physiological studies of synapses in human cells and to pathological studies of diseases that cause abnormal synaptic function. Although it has been reported that synapses themselves change from an immature to a mature state as neurons mature, there are few reports that clearly show when and how human stem cell-derived neurons change to mature synaptic structures. This study was designed to elucidate the synapse formation process of hiPSC-neurons. We propagated hiPSC-derived neural progenitor cells (hiPSC-NPCs) that expressed localized markers of the ventral hindbrain as neurospheres by dual SMAD inhibition and then differentiated them into hiPSC-neurons in vitro. After 49 days of in vitro differentiation, hiPSC-neurons significantly expressed pre- and postsynaptic markers at both the transcript and protein levels. However, the expression of postsynaptic markers was lower than in normal human or normal rat brain tissues, and immunostaining analysis showed that it was relatively modest and was lower than that of presynaptic markers and that its localization in synaptic structures was insufficient. Neurophysiological analysis using a microelectrode array also revealed that no synaptic activity was generated on hiPSC-neurons at 49 days of differentiation. Analysis of subtype markers by immunostaining revealed that most hiPSC-neurons expressed vesicular glutamate transporter 2 (VGLUT2). The presence or absence of NGF, which is required for the survival of cholinergic neurons, had no effect on their cell fractionation. These results suggest that during the synaptogenesis of hiPSC-neurons, the formation of presynaptic structures is not the only requirement for the formation of postsynaptic structures and that the mRNA expression of postsynaptic markers does not correlate with the formation of their mature structures. Technically, we also confirmed a certain level of robustness and reproducibility of our neuronal differentiation method in a multicenter setting, which will be helpful for future research. Synapse formation with mature postsynaptic structures will remain an interesting issue for stem cell-derived neurons, and the present method can be used to obtain early and stable quality neuronal cultures from hiPSC-NPCs.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis , Animals , Biomarkers , Cell Culture Techniques/methods , Cell Line , Hippocampus/cytology , Humans , Induced Pluripotent Stem Cells/drug effects , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/analysis , Neural Stem Cells/ultrastructure , Neurons/chemistry , Neurons/classification , Neurons/cytology , Neuropeptides/analysis , Presynaptic Terminals/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Reproducibility of Results , Synapses/physiology , Vesicular Glutamate Transport Protein 1/analysis , Vesicular Glutamate Transport Protein 2/analysis
2.
Cell Rep ; 37(4): 109895, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706235

ABSTRACT

Neuroblast division is characterized by asymmetric positioning of the cleavage furrow, resulting in a large difference in size between the future daughter cells. In animal cells, furrow placement and assembly are governed by centralspindlin that accumulates at the equatorial cell cortex of the future cleavage site and at the spindle midzone. In neuroblasts, these two centralspindlin populations are spatially and temporally separated. A leading pool is located at the basal cleavage site and a second pool accumulates at the midzone before traveling to the cleavage site. The cortical centralspindlin population requires peripheral astral microtubules and the chromosome passenger complex for efficient recruitment. Loss of this pool does not prevent cytokinesis but enhances centralspindlin signaling at the midzone, leading to equatorial furrow repositioning and decreased size asymmetry. These data show that basal furrow positioning in neuroblasts results from a competition between different centralspindlin pools in which the cortical pool is dominant.


Subject(s)
Cytokinesis , Microtubules , Neural Stem Cells , Animals , Drosophila melanogaster , Microtubules/genetics , Microtubules/metabolism , Microtubules/ultrastructure , Neural Stem Cells/metabolism , Neural Stem Cells/ultrastructure
3.
Elife ; 102021 09 30.
Article in English | MEDLINE | ID: mdl-34590578

ABSTRACT

Astrocytes are essential cells of the central nervous system, characterized by dynamic relationships with neurons that range from functional metabolic interactions and regulation of neuronal firing activities, to the release of neurotrophic and neuroprotective factors. In Parkinson's disease (PD), dopaminergic neurons are progressively lost during the course of the disease, but the effects of PD on astrocytes and astrocyte-to-neuron communication remain largely unknown. This study focuses on the effects of the PD-related mutation LRRK2 G2019S in astrocytes generated from patient-derived induced pluripotent stem cells. We report the alteration of extracellular vesicle (EV) biogenesis in astrocytes and identify the abnormal accumulation of key PD-related proteins within multivesicular bodies (MVBs). We found that dopaminergic neurons internalize astrocyte-secreted EVs and that LRRK2 G2019S EVs are abnormally enriched in neurites and fail to provide full neurotrophic support to dopaminergic neurons. Thus, dysfunctional astrocyte-to-neuron communication via altered EV biological properties may participate in the progression of PD.


Subject(s)
Astrocytes/enzymology , Cell Communication , Dopaminergic Neurons/enzymology , Exosomes/enzymology , Induced Pluripotent Stem Cells/enzymology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neural Stem Cells/enzymology , Parkinson Disease/enzymology , Animals , Astrocytes/ultrastructure , Atrophy , Case-Control Studies , Cell Line , Dopaminergic Neurons/pathology , Endocytosis , Exosomes/genetics , Exosomes/ultrastructure , Humans , Induced Pluripotent Stem Cells/ultrastructure , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neural Stem Cells/ultrastructure , Organelle Biogenesis , Parkinson Disease/genetics , Parkinson Disease/pathology
4.
Sci Rep ; 11(1): 18819, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552130

ABSTRACT

Nanostructured cell culture substrates featuring nanowire (NW) arrays have been applied to a variety of basic cell lines and rodent neurons to investigate cellular behavior or to stimulate cell responses. However, patient-derived human neurons-a prerequisite for studying e.g. neurodegenerative diseases efficiently-are rarely employed due to sensitive cell culture protocols and usually long culturing periods. Here, we present human patient induced pluripotent stem cell-derived neurons cultured on densely-spaced spiky silicon NW arrays (600 NWs/ 100 µm[Formula: see text] with NW lengths of 1 µm) which show mature electrophysiological characteristics after only 20 days of culturing. Exemplary neuronal growth and network formation on the NW arrays are demonstrated using scanning electron microscopy and immunofluorescence microscopy. The cells and neurites rest in a fakir-like settling state on the NWs only in contact with the very NW tips shown by cross-sectional imaging of the cell/NW interface using focused ion beam milling and confocal laser scanning microscopy. Furthermore, the NW arrays promote the cell culture by slightly increasing the share of differentiated neurons determined by the quantification of immunofluorescence microscopy images. The electrophysiological functionality of the neurons is confirmed with patch-clamp recordings showing the excellent capability to fire action potentials. We believe that the short culturing time to obtain functional human neurons generated from patient-derived neural progenitor cells and the robustness of this differentiation protocol to produce these neurons on densely-spaced spiky nanowire arrays open up new pathways for stem cell characterization and neurodegenerative disease studies.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Nanowires , Neural Stem Cells/physiology , Silicon , Humans , Induced Pluripotent Stem Cells/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Neural Stem Cells/ultrastructure
5.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34237032

ABSTRACT

Congenital microcephaly (MCPH) is a neurodevelopmental disease associated with mutations in genes encoding proteins involved in centrosomal and chromosomal dynamics during mitosis. Detailed MCPH pathogenesis at the cellular level is still elusive, given the diversity of MCPH genes and lack of comparative in vivo studies. By generating a series of CRISPR/Cas9-mediated genetic KOs, we report here that - whereas defects in spindle pole proteins (ASPM, MCPH5) result in mild MCPH during development - lack of centrosome (CDK5RAP2, MCPH3) or centriole (CEP135, MCPH8) regulators induces delayed chromosome segregation and chromosomal instability in neural progenitors (NPs). Our mouse model of MCPH8 suggests that loss of CEP135 results in centriole duplication defects, TP53 activation, and cell death of NPs. Trp53 ablation in a Cep135-deficient background prevents cell death but not MCPH, and it leads to subcortical heterotopias, a malformation seen in MCPH8 patients. These results suggest that MCPH in some MCPH patients can arise from the lack of adaptation to centriole defects in NPs and may lead to architectural defects if chromosomally unstable cells are not eliminated during brain development.


Subject(s)
Centrioles/genetics , Chromosomal Instability , Microcephaly/genetics , Neural Stem Cells/pathology , Animals , Brain/cytology , Brain/pathology , CRISPR-Cas Systems/genetics , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrioles/pathology , Disease Models, Animal , Embryo, Mammalian , Female , Humans , Male , Mice , Mice, Knockout , Microcephaly/pathology , Microscopy, Electron, Transmission , Molecular Imaging , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/ultrastructure , Primary Cell Culture , Time-Lapse Imaging , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Int J Mol Sci ; 22(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34074027

ABSTRACT

The development of a biomimetic neuronal network from neural cells is a big challenge for researchers. Recent advances in nanotechnology, on the other hand, have enabled unprecedented tools and techniques for guiding and directing neural stem cell proliferation and differentiation in vitro to construct an in vivo-like neuronal network. Nanotechnology allows control over neural stem cells by means of scaffolds that guide neurons to reform synaptic networks in suitable directions in 3D architecture, surface modification/nanopatterning to decide cell fate and stimulate/record signals from neurons to find out the relationships between neuronal circuit connectivity and their pathophysiological functions. Overall, nanotechnology-mediated methods facilitate precise physiochemical controls essential to develop tools appropriate for applications in neuroscience. This review emphasizes the newest applications of nanotechnology for examining central nervous system (CNS) roles and, therefore, provides an insight into how these technologies can be tested in vitro before being used in preclinical and clinical research and their potential role in regenerative medicine and tissue engineering.


Subject(s)
Cell Culture Techniques/methods , Nanotechnology/methods , Nerve Net/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Tissue Engineering/methods , Animals , Cell Culture Techniques/instrumentation , Humans , Nanotechnology/instrumentation , Nerve Net/ultrastructure , Neural Stem Cells/ultrastructure , Neurogenesis/physiology , Regenerative Medicine , Tissue Engineering/instrumentation
7.
PLoS Biol ; 19(4): e3001166, 2021 04.
Article in English | MEDLINE | ID: mdl-33826607

ABSTRACT

Neural stem cell (NSC) transplantation induces recovery in animal models of central nervous system (CNS) diseases. Although the replacement of lost endogenous cells was originally proposed as the primary healing mechanism of NSC grafts, it is now clear that transplanted NSCs operate via multiple mechanisms, including the horizontal exchange of therapeutic cargoes to host cells via extracellular vesicles (EVs). EVs are membrane particles trafficking nucleic acids, proteins, metabolites and metabolic enzymes, lipids, and entire organelles. However, the function and the contribution of these cargoes to the broad therapeutic effects of NSCs are yet to be fully understood. Mitochondrial dysfunction is an established feature of several inflammatory and degenerative CNS disorders, most of which are potentially treatable with exogenous stem cell therapeutics. Herein, we investigated the hypothesis that NSCs release and traffic functional mitochondria via EVs to restore mitochondrial function in target cells. Untargeted proteomics revealed a significant enrichment of mitochondrial proteins spontaneously released by NSCs in EVs. Morphological and functional analyses confirmed the presence of ultrastructurally intact mitochondria within EVs with conserved membrane potential and respiration. We found that the transfer of these mitochondria from EVs to mtDNA-deficient L929 Rho0 cells rescued mitochondrial function and increased Rho0 cell survival. Furthermore, the incorporation of mitochondria from EVs into inflammatory mononuclear phagocytes restored normal mitochondrial dynamics and cellular metabolism and reduced the expression of pro-inflammatory markers in target cells. When transplanted in an animal model of multiple sclerosis, exogenous NSCs actively transferred mitochondria to mononuclear phagocytes and induced a significant amelioration of clinical deficits. Our data provide the first evidence that NSCs deliver functional mitochondria to target cells via EVs, paving the way for the development of novel (a)cellular approaches aimed at restoring mitochondrial dysfunction not only in multiple sclerosis, but also in degenerative neurological diseases.


Subject(s)
Extracellular Vesicles/metabolism , Mitochondria/metabolism , Neural Stem Cells/metabolism , Animals , Biological Transport , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/ultrastructure
8.
J Neurosci ; 41(19): 4172-4186, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33785644

ABSTRACT

Microglia, the resident immune cells of the CNS, have emerged as key regulators of neural precursor cell activity in the adult brain. However, the microglia-derived factors that mediate these effects remain largely unknown. In the present study, we investigated a role for microglial brain-derived neurotrophic factor (BDNF), a neurotrophic factor with well known effects on neuronal survival and plasticity. Surprisingly, we found that selective genetic ablation of BDNF from microglia increased the production of newborn neurons under both physiological and inflammatory conditions (e.g., LPS-induced infection and traumatic brain injury). Genetic ablation of BDNF from microglia otherwise also interfered with self-renewal/proliferation, reducing their overall density. In conclusion, we identify microglial BDNF as an important factor regulating microglia population dynamics and states, which in turn influences neurogenesis under both homeostatic and pathologic conditions.SIGNIFICANCE STATEMENT (1) Microglial BDNF contributes to self-renewal and density of microglia in the brain. (2) Selective ablation of BDNF in microglia stimulates neural precursor proliferation. (3) Loss of microglial BDNF augments working memory following traumatic brain injury. (4) Benefits of repopulating microglia on brain injury are not mediated via microglial BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Hippocampus/physiology , Microglia/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Animals , Cell Proliferation , Cell Survival/genetics , Dendrites/ultrastructure , Dendritic Spines/ultrastructure , Encephalitis/chemically induced , Encephalitis/pathology , Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/physiology , Neural Stem Cells/ultrastructure
9.
Methods Mol Biol ; 2258: 151-169, 2021.
Article in English | MEDLINE | ID: mdl-33340360

ABSTRACT

Self-assembling brain spheroids derived from human stem cells closely emulate the tangled connectivity of the human brain, recapitulate aspects of organized tissue structure, and are relatively easy to manipulate compared to other existing three-dimensional (3D) cellular models. However, current platforms generate heterogeneously sized and short-lived spheroids, which do not robustly and reproducibly model human brain development and diseases. Here, we present a method to generate large-scale arrays of homogeneously sized 3D brain spheroids derived from human-induced pluripotent stem cells (hiPSCs) or immortalized neural progenitor cells to recapitulate Alzheimer's disease (AD) pathology in vitro. When embedded in extracellular matrix, these brain spheroids develop extensive outward projection of neurites and form networks, which are mediated by thick bundles of dendrites. This array facilitates cost-effective, high-throughput drug screening and mechanistic studies to better understand human brain development and neurodegenerative conditions, such as AD .


Subject(s)
Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Neurogenesis , Neurons/physiology , Tissue Engineering , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Culture Techniques , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Microscopy, Electron, Scanning , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , Neurogenesis/drug effects , Neurons/drug effects , Neurons/ultrastructure , Organoids
10.
Sci Rep ; 10(1): 6596, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313130

ABSTRACT

Enhancement of endogenous neurogenesis after ischemic stroke may improve functional recovery. We previously demonstrated that medium B, which is a combination with epidermal growth factor (EGF) and fibronectin, can promote neural stem/progenitor cell (NSPC) proliferation and migration. Here, we showed that medium B promoted proliferation and migration of cultured NSPCs onto various 3-dimentional structures. When rat cortical neurons with oxygen glucose deprivation (OGD) were co-cultured with NSPCs, medium B treatment increased neuronal viability and reduced cell apoptosis. In a rat model with transient middle cerebral artery occlusion (MCAO), post-insult intraventricular medium B treatment enhanced proliferation, migration, and neuronal differentiation of NSPCs and diminished cell apoptosis in the infarct brain. In cultured post-OGD neuronal cells and the infarct brain from MCAO rats, medium B treatment increased protein levels of Bcl-xL, Bcl-2, phospho-Akt, phospho-GSK-3ß, and ß-catenin and decreased the cleaved caspase-3 level, which may be associated with the effects of anti-apoptosis. Notably, intraventricular medium B treatment increased neuronal density, improved motor function and reduced infarct size in MCAO rats. In summary, medium B treatment results in less neuronal death and better functional outcome in both cellular and rodent models of ischemic stroke, probably via promotion of neurogenesis and reduction of apoptosis.


Subject(s)
Apoptosis , Brain Ischemia/drug therapy , Cerebral Ventricles/pathology , Epidermal Growth Factor/therapeutic use , Fibronectins/therapeutic use , Neurogenesis , Stroke/drug therapy , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Brain Ischemia/physiopathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cerebral Ventricles/physiopathology , Disease Models, Animal , Epidermal Growth Factor/pharmacology , Fibronectins/pharmacology , Glucose/deficiency , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Lateral Ventricles/pathology , Lateral Ventricles/physiopathology , Male , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , Neurogenesis/drug effects , Neurons/drug effects , Neurons/pathology , Oxygen , Rats, Wistar , Recovery of Function/drug effects , Stroke/complications , Stroke/physiopathology
11.
Int J Mol Sci ; 21(5)2020 Mar 02.
Article in English | MEDLINE | ID: mdl-32131546

ABSTRACT

The size of the synaptic subcomponents falls below the limits of visible light microscopy. Despite new developments in advanced microscopy techniques, the resolution of transmission electron microscopy (TEM) remains unsurpassed. The requirements of tissue preservation are very high, and human post mortem material often does not offer adequate quality. However, new reprogramming techniques that generate human neurons in vitro provide samples that can easily fulfill these requirements. The objective of this study was to identify the culture technique with the best ultrastructural preservation in combination with the best embedding and contrasting technique for visualizing neuronal elements. Two induced neural stem cell lines derived from healthy control subjects underwent differentiation either adherent on glass coverslips, embedded in a droplet of highly concentrated Matrigel, or as a compact neurosphere. Afterward, they were fixed using a combination of glutaraldehyde (GA) and paraformaldehyde (PFA) followed by three approaches (standard stain, Ruthenium red stain, high contrast en-bloc stain) using different combinations of membrane enhancing and contrasting steps before ultrathin sectioning and imaging by TEM. The compact free-floating neurospheres exhibited the best ultrastructural preservation. High-contrast en-bloc stain offered particularly sharp staining of membrane structures and the highest quality visualization of neuronal structures. In conclusion, compact neurospheres growing under free-floating conditions in combination with a high contrast en-bloc staining protocol, offer the optimal preservation and contrast with a particular focus on visualizing membrane structures as required for analyzing synaptic structures.


Subject(s)
Cellular Reprogramming Techniques/methods , Microscopy, Electron, Transmission/methods , Neural Stem Cells/ultrastructure , Synapses/ultrastructure , Cells, Cultured , Humans , Neurogenesis
12.
Int J Neurosci ; 130(11): 1101-1108, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32031459

ABSTRACT

Introduction: The transdifferentiation potential of mesenchymal stem cells (MSCs) is not limited to mesodermal derivatives but also to other cell types such as neuronal cells under appropriate cell culture conditions.Materials and methods: The present study characterizes the differentiation of Wharton's jelly (WJ) derived MSCs using neuronal conditioned medium (NCM) collected from cultured foetal brain cells.Results: After induction with NCM to neuronal stem cells (NSC), the WJ MSCs showed profound morphological changes showing multiple neurites extending from the cell body containing reminiscent of Nissl substance and single long axon-like processes. In RT PCR and immunocytochemistry, the induced neuronal cells showed a strong positive expression of neuronal markers Nestin, ß III tubulin and GFAP indicated that, the cells were reactive to NCM for differentiation. A significant (p < 0.01) increase in the level of secretome BDNF was observed in NCM suggests that the BDNF could play a key role in the transdifferentiation of WJMSCs to NSCs.Conclusion: These results support the potential of ovine MSCs isolated from umbilical cord WJ of abattoir derived foetuses to differentiate into neuronal stem cells and also provide a valuable experimental data for NSC transplant research in veterinary medicine.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Transdifferentiation/physiology , Mesenchymal Stem Cells/physiology , Neural Stem Cells/physiology , Wharton Jelly , Animals , Culture Media, Conditioned , Embryo, Mammalian , Glial Fibrillary Acidic Protein/metabolism , Nestin/metabolism , Neural Stem Cells/ultrastructure , Neurites/ultrastructure , Sheep , Tubulin/metabolism , Umbilical Cord , Wharton Jelly/cytology
13.
Epilepsy Behav ; 101(Pt B): 106581, 2019 12.
Article in English | MEDLINE | ID: mdl-31761686

ABSTRACT

Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder caused by deletions in the TSC1 or TSC2 genes that is associated with epilepsy in up to 90% of patients. Seizures are suggested to start in benign brain tumors, cortical tubers, or in the perituberal tissue making these tubers an interesting target for further research into mechanisms underlying epileptogenesis in TSC. Animal models of TSC insufficiently capture the neurodevelopmental biology of cortical tubers, and hence, human stem cell-based in vitro models of TSC are being increasingly explored in attempts to recapitulate tuber development and epileptogenesis in TSC. However, in vitro culture conditions for stem cell-derived neurons do not necessarily mimic physiological conditions. For example, very high glucose concentrations of up to 25 mM are common in culture media formulations. As TSC is potentially caused by a disruption of the mechanistic target of rapamycin (mTOR) pathway, a main integrator of metabolic information and intracellular signaling, we aimed to examine the impact of different glucose concentrations in the culture media on cellular phenotypes implicated in tuber characteristics. Here, we present preliminary data from a pilot study exploring cortical neuronal differentiation on human embryonic stem cells (hES) harboring a TSC2 knockout mutation (TSC2-/-) and an isogenic control line (TSC2+/+). We show that the commonly used high glucose media profoundly mask cellular phenotypes in TSC2-/- cultures during neuronal differentiation. These phenotypes only become apparent when differentiating TSC2+/+ and TSC2-/- cultures in more physiologically relevant conditions of 5 mM glucose suggesting that the careful consideration of culture conditions is vital to ensuring biological relevance and translatability of stem cell models for neurological disorders such as TSC. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures".


Subject(s)
Glucose/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , Tuberous Sclerosis/pathology , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/ultrastructure , Gene Knockout Techniques , Humans , Models, Neurological , Mutation/drug effects , Neurogenesis , Phenotype , Pilot Projects , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics
14.
J Neurosci ; 39(50): 9967-9988, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31685650

ABSTRACT

New neurons, referred to as neuroblasts, are continuously generated in the ventricular-subventricular zone of the brain throughout an animal's life. These neuroblasts are characterized by their unique potential for proliferation, formation of chain-like cell aggregates, and long-distance and high-speed migration through the rostral migratory stream (RMS) toward the olfactory bulb (OB), where they decelerate and differentiate into mature interneurons. The dynamic changes of ultrastructural features in postnatal-born neuroblasts during migration are not yet fully understood. Here we report the presence of a primary cilium, and its ultrastructural morphology and spatiotemporal dynamics, in migrating neuroblasts in the postnatal RMS and OB. The primary cilium was observed in migrating neuroblasts in the postnatal RMS and OB in male and female mice and zebrafish, and a male rhesus monkey. Inhibition of intraflagellar transport molecules in migrating neuroblasts impaired their ciliogenesis and rostral migration toward the OB. Serial section transmission electron microscopy revealed that each migrating neuroblast possesses either a pair of centrioles or a basal body with an immature or mature primary cilium. Using immunohistochemistry, live imaging, and serial block-face scanning electron microscopy, we demonstrate that the localization and orientation of the primary cilium are altered depending on the mitotic state, saltatory migration, and deceleration of neuroblasts. Together, our results highlight a close mutual relationship between spatiotemporal regulation of the primary cilium and efficient chain migration of neuroblasts in the postnatal brain.SIGNIFICANCE STATEMENT Immature neurons (neuroblasts) generated in the postnatal brain have a mitotic potential and migrate in chain-like cell aggregates toward the olfactory bulb. Here we report that migrating neuroblasts possess a tiny cellular protrusion called a primary cilium. Immunohistochemical studies with zebrafish, mouse, and monkey brains suggest that the presence of the primary cilium in migrating neuroblasts is evolutionarily conserved. Ciliogenesis in migrating neuroblasts in the rostral migratory stream is suppressed during mitosis and promoted after cell cycle exit. Moreover, live imaging and 3D electron microscopy revealed that ciliary localization and orientation change during saltatory movement of neuroblasts. Our results reveal highly organized dynamics in maturation and positioning of the primary cilium during neuroblast migration that underlie saltatory movement of postnatal-born neuroblasts.


Subject(s)
Cell Movement/physiology , Cilia/ultrastructure , Lateral Ventricles/ultrastructure , Neural Stem Cells/ultrastructure , Neurons/ultrastructure , Olfactory Bulb/ultrastructure , Animals , Female , Macaca mulatta , Male , Mice , Zebrafish
15.
Mater Sci Eng C Mater Biol Appl ; 102: 34-44, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31147006

ABSTRACT

Spinal cord injury (SCI) is one of the most precarious conditions which have been one of the major reasons for continuous increasing mortality rate of SCI patients. Currently, there is no effective treatment modality for SCI patients posing major threat to the scientific and medical community. The available strategies don't mimic with the natural processes of nervous tissues repair/regeneration and majority of the approaches may induce the additional fibrotic or immunological response at the injury site and are not readily available on demand. To overcome these hurdles, we have developed a ready to use bioengineered human functional neurological construct (BHNC) for regenerative applications in SCI defects. We used cryopreserved meningeal tissues (CMT) for bioengineering these neurological constructs using acellularization and repopulation technology. The technology adopted herein generates intact neurological scaffolds from CMT and retains several crucial structural, biochemical and mechanical cues to enhance the regenerative mechanisms. The neurogenic differentiation on CMT scaffolds was almost similar to the freshly prepared meningeal scaffolds and mimics with the natural nervous tissue developmental mechanisms which offer intact 3D-microarchitecture and hospitable microenvironment enriched with several crucial neurotrophins for long-term cell survival and function. Functional assessment of developed BHNC showed highly increased positive staining for pre-synaptic granules of Synapsis-1 along with MAP-2 antibody with punctuate distribution in axonal regions of the neuronal cells which was well supported by the gene expression analysis of functional transcripts. Given the significant improvement in the field may enable to generate more such ready to use functional BHNC for wider applicability in SCI repair/regeneration.


Subject(s)
Biomimetic Materials/pharmacology , Cryopreservation , Meninges/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biomechanical Phenomena , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Humans , Meninges/drug effects , Meninges/ultrastructure , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Arch Toxicol ; 93(6): 1585-1608, 2019 06.
Article in English | MEDLINE | ID: mdl-31190196

ABSTRACT

Many neurotoxicants affect energy metabolism in man, but currently available test methods may still fail to predict mito- and neurotoxicity. We addressed this issue using LUHMES cells, i.e., human neuronal precursors that easily differentiate into mature neurons. Within the NeuriTox assay, they have been used to screen for neurotoxicants. Our new approach is based on culturing the cells in either glucose or galactose (Glc-Gal-NeuriTox) as the main carbohydrate source during toxicity testing. Using this Glc-Gal-NeuriTox assay, 52 mitochondrial and non-mitochondrial toxicants were tested. The panel of chemicals comprised 11 inhibitors of mitochondrial respiratory chain complex I (cI), 4 inhibitors of cII, 8 of cIII, and 2 of cIV; 8 toxicants were included as they are assumed to be mitochondrial uncouplers. In galactose, cells became more dependent on mitochondrial function, which made them 2-3 orders of magnitude more sensitive to various mitotoxicants. Moreover, galactose enhanced the specific neurotoxicity (destruction of neurites) compared to a general cytotoxicity (plasma membrane lysis) of the toxicants. The Glc-Gal-NeuriTox assay worked particularly well for inhibitors of cI and cIII, while the toxicity of uncouplers and non-mitochondrial toxicants did not differ significantly upon glucose ↔ galactose exchange. As a secondary assay, we developed a method to quantify the inhibition of all mitochondrial respiratory chain functions/complexes in LUHMES cells. The combination of the Glc-Gal-NeuriTox neurotoxicity screening assay with the mechanistic follow up of target site identification allowed both, a more sensitive detection of neurotoxicants and a sharper definition of the mode of action of mitochondrial toxicants.


Subject(s)
Mitochondria/drug effects , Mitochondrial Diseases/chemically induced , Neural Stem Cells/drug effects , Neurotoxicity Syndromes/diagnosis , Toxicity Tests/methods , Carbohydrate Metabolism , Culture Media , Electron Transport/drug effects , Electron Transport Complex I/antagonists & inhibitors , Galactose/metabolism , Galactose/pharmacology , Glucose/metabolism , Glucose/pharmacology , Humans , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Neural Stem Cells/ultrastructure , Neurites/drug effects , Uncoupling Agents/toxicity
17.
Adv Mater ; 31(32): e1900488, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31197896

ABSTRACT

Volumetric imaging techniques capable of correlating structural and functional information with nanoscale resolution are necessary to broaden the insight into cellular processes within complex biological systems. The recent emergence of focused ion beam scanning electron microscopy (FIB-SEM) has provided unparalleled insight through the volumetric investigation of ultrastructure; however, it does not provide biomolecular information at equivalent resolution. Here, immunogold FIB-SEM, which combines antigen labeling with in situ FIB-SEM imaging, is developed in order to spatially map ultrastructural and biomolecular information simultaneously. This method is applied to investigate two different cell-material systems: the localization of histone epigenetic modifications in neural stem cells cultured on microstructured substrates and the distribution of nuclear pore complexes in myoblasts differentiated on a soft hydrogel surface. Immunogold FIB-SEM offers the potential for broad applicability to correlate structure and function with nanoscale resolution when addressing questions across cell biology, biomaterials, and regenerative medicine.


Subject(s)
Microscopy, Electron, Scanning/methods , Myoblasts/cytology , Neural Stem Cells/ultrastructure , Nuclear Pore/ultrastructure , Cell Differentiation , Dimethylpolysiloxanes , Epigenesis, Genetic , Humans , Hydrogels , Imaging, Three-Dimensional
18.
Commun Biol ; 2: 155, 2019.
Article in English | MEDLINE | ID: mdl-31069265

ABSTRACT

Current approaches for dynamic profiling of single cells rely on dissociated cultures, which lack important biological features existing in tissues. Organotypic slice cultures preserve aspects of structural and synaptic organisation within the brain and are amenable to microscopy, but established techniques are not well adapted for high throughput or longitudinal single cell analysis. Here we developed a custom-built, automated confocal imaging platform, with improved organotypic slice culture and maintenance. The approach enables fully automated image acquisition and four-dimensional tracking of morphological changes within individual cells in organotypic cultures from rodent and human primary tissues for at least 3 weeks. To validate this system, we analysed neurons expressing a disease-associated version of huntingtin (HTT586Q138-EGFP), and observed that they displayed hallmarks of Huntington's disease and died sooner than controls. By facilitating longitudinal single-cell analyses of neuronal physiology, our system bridges scales necessary to attain statistical power to detect developmental and disease phenotypes.


Subject(s)
Cell Tracking/methods , Hippocampus/ultrastructure , Huntington Disease/pathology , Microscopy, Confocal/methods , Neurons/ultrastructure , Single-Cell Analysis/methods , Amino Acid Substitution , Animals , Animals, Newborn , Cell Differentiation , Cell Tracking/instrumentation , Gene Expression , Hippocampus/metabolism , Hippocampus/pathology , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal/instrumentation , Models, Biological , Neural Stem Cells/metabolism , Neural Stem Cells/ultrastructure , Neurons/metabolism , Primary Cell Culture , Single-Cell Analysis/instrumentation , Tissue Culture Techniques
19.
Cell Death Dis ; 10(3): 211, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824686

ABSTRACT

Offspring of mothers with hyperglycemia during pregnancy have a higher incidence of long-term neuropsychiatric disorders than offspring from a normal pregnancy, indicating that neocortical neurogenesis might be affected by maternal hyperglycemia. A paucity of study evaluating the effects of hyperglycemia on neocortical neurogenetic differentiation of neural stem cells, and the mechanism remains unclear. We sought to investigate the the roles and possible molecular mechanism of maternal hyperglycemia on neocortical neurogenetic differentiation of neural stem cells. We established a mouse model of a hyperglycemic pregnancy to study effects of intrauterine exposure to maternal hyperglycemia on neocortical neurogenesis. We observed morphological changes in the neocortex and detected the neurogenetic differentiation of neural stem cells in offspring affected by high glucose levels. We investigated the regulatory network between epigenetic modification and transcription factors in differentiated neural stem cells under hyperglycemic conditions. Maternal hyperglycemia disturbs neocortical lamination in some non-malformed offspring. Our results suggested that hyperglycemia altered the early-born neuron fate and the distribution of newborn neurons in deep layers by promoting the earlier differentiation of neural stem cells. Altered histone acetylation and its regulation on the transcription of proneural genes might be correlated to the disrupted differentiation of neural stem cells and altered distribution of newborn projection neurons in the neocortex. Our data raised the possibility that maternal hyperglycemia in pregnancy disturbs the laminar distribution of neocortical projection neurons in some non-malformed offspring via epigenetic regulation on neural stem cell differentiation and the birthdate of neocortical neurons.


Subject(s)
Epigenesis, Genetic , Hyperglycemia , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , Pregnancy Complications , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/chemistry , Histones/metabolism , Mice , Mice, Inbred C57BL , Neocortex/growth & development , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/ultrastructure , Neural Tube Defects/pathology , Neurons/cytology , Pregnancy
20.
Nat Med ; 25(2): 263-269, 2019 02.
Article in English | MEDLINE | ID: mdl-30643285

ABSTRACT

Current methods for bioprinting functional tissue lack appropriate biofabrication techniques to build complex 3D microarchitectures essential for guiding cell growth and promoting tissue maturation1. 3D printing of central nervous system (CNS) structures has not been accomplished, possibly owing to the complexity of CNS architecture. Here, we report the use of a microscale continuous projection printing method (µCPP) to create a complex CNS structure for regenerative medicine applications in the spinal cord. µCPP can print 3D biomimetic hydrogel scaffolds tailored to the dimensions of the rodent spinal cord in 1.6 s and is scalable to human spinal cord sizes and lesion geometries. We tested the ability of µCPP 3D-printed scaffolds loaded with neural progenitor cells (NPCs) to support axon regeneration and form new 'neural relays' across sites of complete spinal cord injury in vivo in rodents1,2. We find that injured host axons regenerate into 3D biomimetic scaffolds and synapse onto NPCs implanted into the device and that implanted NPCs in turn extend axons out of the scaffold and into the host spinal cord below the injury to restore synaptic transmission and significantly improve functional outcomes. Thus, 3D biomimetic scaffolds offer a means of enhancing CNS regeneration through precision medicine.


Subject(s)
Biomimetics , Nerve Regeneration , Printing, Three-Dimensional , Spinal Cord Injuries/therapy , Tissue Scaffolds/chemistry , Animals , Green Fluorescent Proteins/metabolism , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Neural Stem Cells/ultrastructure , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...